Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Br J Clin Pharmacol ; 89(7): 2190-2200, 2023 07.
Article in English | MEDLINE | ID: mdl-36740580

ABSTRACT

AIM: SWORD-1 and SWORD-2 phase 3 studies concluded that switching virologically suppressed participants with HIV-1 from their current three- or four-drug antiretroviral regimen (CAR) to the two-drug regimen of once-daily dolutegravir (DTG, 50 mg) and rilpivirine (RPV, 25 mg) was safe, well tolerated and noninferior for maintaining HIV-1 suppression at week 48 and highly efficacious to week 148. A secondary objective was to characterize drug exposure and exposure-efficacy/safety relationships. METHODS: Adults with plasma HIV-1 RNA <50 copies/mL were randomized to switch to once-daily DTG + RPV on day 1 or to continue CAR for 52 weeks before switching. Trough plasma concentrations (C0) of DTG and RPV, the proportion of participants with HIV-1 RNA <50 copies/mL and adverse events to week 100 were summarized and subjected to exposure-response analyses in the overall population, in the subset of participants who switched from CAR containing enzyme-inducing drugs and by age category (≥50 and <50 years). The relationship between C0avg (individual average C0 across visits) and efficacy/safety was investigated. RESULTS: Although week 2 DTG and RPV C0 were lower in participants switching from enzyme-inducing antiretroviral drugs, C0 and C0avg stayed above in vitro antiviral protein binding-adjusted IC90 and to week 100 with viral suppression >89%. DTG or RPV C0avg showed no relationship with virologic failures or safety. Participants ≥50 years had similar C0avg and safety response to younger participants. CONCLUSION: No clinically relevant relationship between DTG or RPV exposures and virologic or safety response was observed, confirming the DTG + RPV switch for participants as a safe and effective treatment.


Subject(s)
Anti-HIV Agents , HIV Infections , Adult , Humans , Middle Aged , HIV Infections/drug therapy , Rilpivirine/adverse effects , Oxazines , Pyridones/therapeutic use , Heterocyclic Compounds, 3-Ring/adverse effects , Anti-Retroviral Agents/therapeutic use , Treatment Outcome , RNA , Viral Load
2.
Pediatr Infect Dis J ; 41(3): 230-237, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34817414

ABSTRACT

BACKGROUND: The World Health Organization (WHO) 2019 antiretroviral treatment guidelines recommend use of optimal treatment regimens in all populations. Dolutegravir-based regimens are the preferred first-line and second-line treatment in infants and children with HIV 4 weeks of age and above. There is an urgent need for optimal pediatric formulations of dolutegravir as single-entity (SE) and fixed-dose combination (FDC) to ensure correct dosing and adherence for swallowing and palatability. This article outlines the chronology of dolutegravir pediatric formulation development as granules and conventional and dispersible tablets in a total of 5 pharmacokinetic studies evaluating the relative bioavailability of dolutegravir SE and FDC formulations in healthy adults. METHODS: The relative bioavailability studies were 2-part, Phase I, open-label, randomized studies in healthy adults. Dolutegravir SE study compared conventional dolutegravir 50 and 25 mg with equivalent conventional 10-mg and dispersible 5-mg tablets, respectively. Subsequently, dolutegravir FDC study compared adult FDC of abacavir/dolutegravir/lamivudine and adult FDC of dolutegravir/lamivudine with their respective pediatric FDC formulations, taken as dispersion immediately or swallowed whole. RESULTS: As observed in previous studies, dolutegravir administered as dispersion (granules/dispersible tablets) showed relatively higher bioavailability compared with conventional tablets. The bioavailability of dolutegravir dispersible tablets (both SE and FDC) was approximately 1.6-fold higher when compared with conventional tablets. In addition, the bioavailability of abacavir/lamivudine was not impacted by dispersible formulation. CONCLUSIONS: These studies demonstrate the successful development of pediatric dolutegravir-containing formulations as SE and FDC that permit pediatric dosing in line with WHO recommendations.


Subject(s)
Anti-HIV Agents/administration & dosage , Heterocyclic Compounds, 3-Ring/administration & dosage , Oxazines/administration & dosage , Piperazines/administration & dosage , Pyridones/administration & dosage , Acquired Immunodeficiency Syndrome/drug therapy , Administration, Oral , Adolescent , Adult , Aged , Anti-HIV Agents/pharmacokinetics , Biological Availability , Dideoxynucleosides , Drug Combinations , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Humans , Lamivudine , Middle Aged , Oxazines/pharmacokinetics , Piperazines/pharmacokinetics , Pyridones/pharmacokinetics , Tablets/administration & dosage , Young Adult
3.
J Clin Pharmacol ; 55(9): 1042-50, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25857714

ABSTRACT

JNJ-56914845 (GSK2336805) is a hepatitis C virus nonstructural protein 5A inhibitor under development for the treatment of chronic hepatitis C (CHC) infection. This open-label, parallel-group, 2-part study evaluated the pharmacokinetics and safety of a single oral 60 mg dose of JNJ-56914845 in 4 cohorts: healthy, mild, moderate, and severe hepatic impairment (n = 8/cohort). Severity of hepatic impairment was categorized using Child-Pugh score, and the healthy subjects were matched for age, sex, body mass index, and smoking status to the moderate hepatic impairment cohort. JNJ-56914845 plasma AUC0-∞ was 26%, 52%, and 45% lower in subjects with mild, moderate, and severe hepatic impairment, respectively, relative to healthy subjects with no difference in half-life among the groups. The apparent oral clearance and volume of distribution were higher in subjects with hepatic impairment. The lower plasma concentrations were largely explained by decreased plasma protein binding in hepatically impaired subjects. One subject with severe hepatic impairment had 2 non-drug-related serious adverse events: an esophageal bleed requiring hospitalization, encephalopathy. Although hepatically impaired subjects have lower exposures than healthy matched controls, they had similar or slightly higher exposures than those observed in past studies of noncirrhotic, CHC patients, suggesting that no dose adjustments for hepatic impairment will be needed.


Subject(s)
Antiviral Agents/pharmacokinetics , Carbamates/pharmacokinetics , Hepacivirus/metabolism , Liver Failure/metabolism , Valine/analogs & derivatives , Viral Nonstructural Proteins/antagonists & inhibitors , Adult , Area Under Curve , Carbamates/adverse effects , Female , Half-Life , Humans , Male , Middle Aged , Valine/adverse effects , Valine/pharmacokinetics
4.
Clin Pharmacol Drug Dev ; 3(5): 338-45, 2014 09.
Article in English | MEDLINE | ID: mdl-27129005

ABSTRACT

This Phase I, randomized, open-label study evaluated the gastric pH-altering effects of omeprazole, a proton pump inhibitor, and the CYP3A enzyme/P-glycoprotein (Pgp)-inhibitory effects of ritonavir, an HIV protease inhibitor, on the pharmacokinetics and safety of the hepatitis C virus (HCV) non-structural protein 5A (NS5A) inhibitor GSK2336805 in healthy male and female subjects. Co-administration of GSK2336805 60 mg with omeprazole decreased GSK2336805 plasma AUC(0-∞) by 10% and Cmax by 18%; no marked effect was observed on t½ . Co-administration of GSK2336805 30 mg with ritonavir increased GSK2336805 plasma AUC(0-∞) by 52%, Cmax by 43%, and t½ by 40%; CL/F was decreased by 34%. All adverse events were minor in intensity. The gastric acid-suppressive effect of omeprazole had minimal impact on the extent and rate of GSK2336805 absorption in vivo; therefore, GSK2336805 may be co-administered with omeprazole without concern about lower GSK2336805 exposures and compromised antiviral efficacy. The modest increases in AUC and Cmax following co-administration of GSK2336805 plus ritonavir suggest that GSK2336805 when given concomitantly with a single CYP3A/Pgp inhibiting drug will not likely require dose adjustment. Final dose recommendation will be based on GSK2336805 efficacy and safety profiles from Phase III trials in HCV-infected patients.


Subject(s)
Antiviral Agents/pharmacokinetics , Carbamates/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A/metabolism , Gastrointestinal Absorption , Hepacivirus/drug effects , Omeprazole/administration & dosage , Protease Inhibitors/pharmacokinetics , Proton Pump Inhibitors/administration & dosage , Ritonavir/administration & dosage , Valine/analogs & derivatives , Viral Nonstructural Proteins/antagonists & inhibitors , Adolescent , Adult , Aged , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/blood , Area Under Curve , Biotransformation , Carbamates/administration & dosage , Carbamates/adverse effects , Carbamates/blood , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Drug Interactions , Female , Half-Life , Healthy Volunteers , Hepacivirus/enzymology , Humans , Male , Metabolic Clearance Rate , Middle Aged , New York , Omeprazole/adverse effects , Protease Inhibitors/administration & dosage , Protease Inhibitors/adverse effects , Protease Inhibitors/blood , Proton Pump Inhibitors/adverse effects , Ritonavir/adverse effects , Valine/administration & dosage , Valine/adverse effects , Valine/blood , Valine/pharmacokinetics , Viral Nonstructural Proteins/metabolism , Young Adult
5.
J Pharm Sci ; 100(10): 4127-57, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21541937

ABSTRACT

The objective of this study is to assess the effectiveness of physiologically based pharmacokinetic (PBPK) models for simulating human plasma concentration-time profiles for the unique drug dataset of blinded data that has been assembled as part of a Pharmaceutical Research and Manufacturers of America initiative. Combinations of absorption, distribution, and clearance models were tested with a PBPK approach that has been developed from published equations. An assessment of the quality of the model predictions was made on the basis of the shape of the plasma time courses and related parameters. Up to 69% of the simulations of plasma time courses made in human demonstrated a medium to high degree of accuracy for intravenous pharmacokinetics, whereas this number decreased to 23% after oral administration based on the selected criteria. The simulations resulted in a general underestimation of drug exposure (Cmax and AUC0- t ). The explanations for this underestimation are diverse. Therefore, in general it may be due to underprediction of absorption parameters and/or overprediction of distribution or oral first-pass. The implications of compound properties are demonstrated. The PBPK approach based on in vitro-input data was as accurate as the approach based on in vivo data. Overall, the scientific benefit of this modeling study was to obtain more extensive characterization of predictions of human PK from PBPK methods.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Administration, Oral , Animals , Computer Simulation , Cooperative Behavior , Drug Evaluation, Preclinical , Gastrointestinal Absorption , Humans , Interdisciplinary Communication , Metabolic Clearance Rate , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Program Development , Program Evaluation , Reproducibility of Results , Species Specificity
6.
J Pharm Sci ; 100(10): 4090-110, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21541938

ABSTRACT

The objective of this study was to evaluate the performance of various allometric and in vitro-in vivo extrapolation (IVIVE) methodologies with and without plasma protein binding corrections for the prediction of human intravenous (i.v.) clearance (CL). The objective was also to evaluate the IVIVE prediction methods with animal data. Methodologies were selected from the literature. Pharmaceutical Research and Manufacturers of America member companies contributed blinded datasets from preclinical and clinical studies for 108 compounds, among which 19 drugs had i.v. clinical pharmacokinetics data and were used in the analysis. In vivo and in vitro preclinical data were used to predict CL by 29 different methods. For many compounds, in vivo data from only two species (generally rat and dog) were available and/or the required in vitro data were missing, which meant some methods could not be properly evaluated. In addition, 66 methods of predicting oral (p.o.) area under the curve (AUCp.o. ) were evaluated for 107 compounds using rational combinations of i.v. CL and bioavailability (F), and direct scaling of observed p.o. CL from preclinical species. Various statistical and outlier techniques were employed to assess the predictability of each method. Across methods, the maximum success rate in predicting human CL for the 19 drugs was 100%, 94%, and 78% of the compounds with predictions falling within 10-fold, threefold, and twofold error, respectively, of the observed CL. In general, in vivo methods performed slightly better than IVIVE methods (at least in terms of measures of correlation and global concordance), with the fu intercept method and two-species-based allometry (rat-dog) being the best performing methods. IVIVE methods using microsomes (incorporating both plasma and microsomal binding) and hepatocytes (not incorporating binding) resulted in 75% and 78%, respectively, of the predictions falling within twofold error. IVIVE methods using other combinations of binding assumptions were much less accurate. The results for prediction of AUCp.o. were consistent with i.v. CL. However, the greatest challenge to successful prediction of human p.o. CL is the estimate of F in human. Overall, the results of this initiative confirmed predictive performance of common methodologies used to predict human CL.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Animals , Area Under Curve , Computer Simulation , Cooperative Behavior , Dogs , Drug Evaluation, Preclinical , Humans , Interdisciplinary Communication , Metabolic Clearance Rate , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Program Development , Program Evaluation , Protein Binding , Rats , Reproducibility of Results , Species Specificity
7.
J Pharm Sci ; 100(10): 4050-73, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21523782

ABSTRACT

This study is part of the Pharmaceutical Research and Manufacturers of America (PhRMA) initiative on predictive models of efficacy, safety, and compound properties. The overall goal of this part was to assess the predictability of human pharmacokinetics (PK) from preclinical data and to provide comparisons of available prediction methods from the literature, as appropriate, using a representative blinded dataset of drug candidates. The key objectives were to (i) appropriately assemble and blind a diverse dataset of in vitro, preclinical in vivo, and clinical data for multiple drug candidates, (ii) evaluate the dataset with empirical and physiological methodologies from the literature used to predict human PK properties and plasma concentration-time profiles, (iii) compare the predicted properties with the observed clinical data to assess the prediction accuracy using routine statistical techniques and to evaluate prediction method(s) based on the degree of accuracy of each prediction method, and (iv) compile and summarize results for publication. Another objective was to provide a mechanistic understanding as to why one methodology provided better predictions than another, after analyzing the poor predictions. A total of 108 clinical lead compounds were collected from 12 PhRMA member companies. This dataset contains intravenous (n = 19) and oral pharmacokinetic data (n = 107) in humans as well as the corresponding preclinical in vitro, in vivo, and physicochemical data. All data were blinded to protect the anonymity of both the data and the company submitting the data. This manuscript, which is the first of a series of manuscripts, summarizes the PhRMA initiative and the 108 compound dataset. More details on the predictability of each method are reported in companion manuscripts.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Administration, Oral , Animals , Computer Simulation , Cooperative Behavior , Drug Evaluation, Preclinical , Humans , Interdisciplinary Communication , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/chemistry , Program Development , Program Evaluation , Reproducibility of Results , Risk Assessment , Risk Factors , Species Specificity
8.
J Pharm Sci ; 100(10): 4074-89, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21452299

ABSTRACT

The objective of this study was to evaluate the performance of various empirical, semimechanistic and mechanistic methodologies with and without protein binding corrections for the prediction of human volume of distribution at steady state (Vss ). PhRMA member companies contributed a set of blinded data from preclinical and clinical studies, and 18 drugs with intravenous clinical pharmacokinetics (PK) data were available for the analysis. In vivo and in vitro preclinical data were used to predict Vss by 24 different methods. Various statistical and outlier techniques were employed to assess the predictability of each method. There was not simply one method that predicts Vss accurately for all compounds. Across methods, the maximum success rate in predicting human Vss was 100%, 94%, and 78% of the compounds with predictions falling within tenfold, threefold, and twofold error, respectively, of the observed Vss . Generally, the methods that made use of in vivo preclinical data were more predictive than those methods that relied solely on in vitro data. However, for many compounds, in vivo data from only two species (generally rat and dog) were available and/or the required in vitro data were missing, which meant some methods could not be properly evaluated. It is recommended to initially use the in vitro tissue composition-based equations to predict Vss in preclinical species and humans, putting the assumptions and compound properties into context. As in vivo data become available, these predictions should be reassessed and rationalized to indicate the level of confidence (uncertainty) in the human Vss prediction. The top three methods that perform strongly at integrating in vivo data in this way were the Øie-Tozer, the rat -dog-human proportionality equation, and the lumped-PBPK approach. Overall, the scientific benefit of this study was to obtain greater characterization of predictions of human Vss from several methods available in the literature.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Animals , Computer Simulation , Cooperative Behavior , Dogs , Drug Evaluation, Preclinical , Humans , Interdisciplinary Communication , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Program Development , Program Evaluation , Protein Binding , Rats , Reproducibility of Results , Species Specificity
9.
J Pharm Sci ; 100(10): 4111-26, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21480234

ABSTRACT

The objective of this study was to evaluate the performance of the Wajima allometry (Css -MRT) approach published in the literature, which is used to predict the human plasma concentration-time profiles from a scaling of preclinical species data. A diverse and blinded dataset of 108 compounds from PhRMA member companies was used in this evaluation. The human intravenous (i.v.) and oral (p.o.) pharmacokinetics (PK) data were available for 18 and 107 drugs, respectively. Three different scenarios were adopted for prediction of human PK profiles. In the first scenario, human clearance (CL) and steady-state volume of distribution (Vss ) were predicted by unbound fraction corrected intercept method (FCIM) and Øie-Tozer (OT) approaches, respectively. Quantitative structure activity relationship (QSAR)-based approaches (TSrat-dog ) based on compound descriptors together with rat and dog data were utilized in the second scenario. Finally, in the third scenario, CL and Vss were predicted using the FCIM and Jansson approaches, respectively. For the prediction of oral pharmacokinetics, the human bioavailability and absorption rate constant were assumed as the average of preclinical species. Various statistical techniques were used for assessing the accuracy of the simulation scenarios. The human CL and Vss were predicted within a threefold error range for about 75% of the i.v. drugs. However, the accuracy in predicting key p.o. PK parameters appeared to be lower with only 58% of simulations falling within threefold of observed parameters. The overall ability of the Css -MRT approach to predict the curve shape of the profile was in general poor and ranged between low to medium level of confidence for most of the predictions based on the selected criteria.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Administration, Oral , Animals , Biological Availability , Computer Simulation , Cooperative Behavior , Dogs , Drug Evaluation, Preclinical , Gastrointestinal Absorption , Humans , Interdisciplinary Communication , Metabolic Clearance Rate , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Program Development , Program Evaluation , Rats , Reproducibility of Results , Species Specificity
10.
J Pharm Sci ; 99(2): 1046-62, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19569219

ABSTRACT

This study evaluated the utility of oral sulfasalazine as a probe substrate for Breast Cancer Resistance Protein (BCRP; ABCG2) activity by assessing the impact of genetic variation or coadministration of an inhibitor (pantoprazole) on plasma and urine pharmacokinetics of sulfasalazine and metabolites. Thirty-six healthy male subjects prescreened for ABCG2 421CC (reference activity), CA, and AA (lower activity) genotypes (N = 12 each) received a single 500 mg oral dose of enteric coated sulfasalazine alone, with 40 mg pantoprazole, or with 40 mg famotidine (gastrointestinal pH control) in a 3-period, single fixed sequence, crossover design. No significant difference in sulfasalazine or metabolite pharmacokinetics in 421AA or CA compared to 421CC subjects was found; however, high inter-subject variability was observed. Geometric mean (95% CI) sulfasalazine plasma AUC((0-infinity)) values were 32.1 (13.2, 78.1), 16.8 (7.15, 39.6) and 62.7 (33.4, 118) microg h/mL, and C(max) were 4.01 (1.62, 9.92), 1.70 (0.66, 4.40), and 6.86 (3.61, 13.0) microg/mL for CC, CA, and AA subjects, respectively. Pantoprazole and famotidine did not affect sulfasalazine pharmacokinetics in any genotypic cohort. These results suggest that the pharmacokinetics of oral, enteric-coated 500 mg sulfasalazine are not sufficiently sensitive to ABCG2 genetic variation or inhibitors to be useful as a clinical probe substrate of BCRP activity.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/pharmacokinetics , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Enzyme Inhibitors/pharmacokinetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Sulfasalazine/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Adult , Arylamine N-Acetyltransferase/genetics , Arylamine N-Acetyltransferase/metabolism , Biotransformation , Chromatography, High Pressure Liquid , Cross-Over Studies , Famotidine/pharmacokinetics , Gastric Mucosa/metabolism , Genotype , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Neoplasm Proteins/metabolism , Pantoprazole , Pharmacogenetics , Polymorphism, Genetic , Proton Pump Inhibitors/pharmacokinetics , Spectrophotometry, Ultraviolet , Young Adult
11.
J Clin Pharmacol ; 48(10): 1179-88, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18676693

ABSTRACT

Aplaviroc (GW873140) binds specifically to human cellular CC chemokine receptor 5 (CCR5) and demonstrates potent anti-human immunodeficiency virus activity in vitro in the subnanomolar range. In vitro studies show that aplaviroc selectively inhibits the binding of a particular monoclonal antibody, 45531, to CCR5. Based on this observation, a flow cytometry-based assay was developed to determine percentage CCR5 receptor occupancy (RO). CCR5 receptor occupancy was aplaviroc concentration-dependent and related to anti-human immunodeficiency virus activity in vitro. In the clinical setting, CCR5 receptor occupancy in peripheral blood was >98% in all subjects within 2 to 3 hours of dosing, which is consistent with the peak plasma concentrations of drug. Longitudinal analysis in the drug washout period revealed the time to 50% CCR5 receptor occupancy averaged >100 hours, in both human immunodeficiency virus-positive and human immunodeficiency virus-negative subjects, substantially longer than the plasma pharmacokinetic half-life of 3 hours. The duration of CCR5 receptor occupancy appeared to be dose-dependent and associated with antiviral activity as measured by plasma human immunodeficiency virus RNA nadir following 10 days of multiple dose administration. These data demonstrate that the analysis of CCR5 receptor occupancy, in addition to conventional plasma-based pharmacokinetic measures, provides an informative tool to assist in evaluating the pharmacodynamic and antiviral effects of cellular CC chemokine receptor antagonists.


Subject(s)
Benzoates/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV Infections/drug therapy , Piperazines/pharmacology , Receptors, CCR5/drug effects , Spiro Compounds/pharmacology , Adult , Antibodies, Monoclonal/metabolism , Benzoates/administration & dosage , Benzoates/pharmacokinetics , Diketopiperazines , Dose-Response Relationship, Drug , Double-Blind Method , Female , Flow Cytometry , HIV Fusion Inhibitors/administration & dosage , HIV Fusion Inhibitors/pharmacokinetics , Half-Life , Humans , Male , Middle Aged , Piperazines/administration & dosage , Piperazines/pharmacokinetics , Protein Binding/drug effects , RNA, Viral/blood , RNA, Viral/drug effects , Receptors, CCR5/metabolism , Spiro Compounds/administration & dosage , Spiro Compounds/pharmacokinetics , Time Factors , Young Adult
12.
Br J Clin Pharmacol ; 66(2): 233-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18429968

ABSTRACT

AIMS: A number of drugs are substrates or inhibitors of the efflux transporter breast cancer resistance protein (BCRP; ABCG2), which can limit systemic exposure by reducing absorption and/or increasing biliary elimination. The identification of a BCRP-selective clinical probe drug would provide a useful tool to understand the effect of genetic polymorphisms and transporter-based drug interactions on drug pharmacokinetics. The aim of this study was to assess the utility of nitrofurantoin as a clinical probe substrate for BCRP activity by evaluating the impact of genetic variation on nitrofurantoin pharmacokinetics. METHODS: Nitrofurantoin pharmacokinetics were studied in an open-label, single-oral dose (100 mg) study in 36 male Chinese subjects who were pre-screened for ABCG2 421 CC, CA and AA genotypes (n = 12 each). Plasma and urine concentrations of nitrofurantoin were determined by LC/MS/MS and LC/UV respectively. anova was used to compare pharmacokinetic parameters among genotypes. RESULTS: There were no significant differences in nitrofurantoin pharmacokinetics among the genotypic cohorts. The geometric mean nitrofurantoin plasma AUC((0-infinity)) (95% confidence interval) values were 2.21 (2.00, 2.45), 2.42 (2.11, 2.78) and 2.32 (1.99, 2.70) microg h ml(-1) and half-life values were 0.79 (0.59, 1.0), 0.76 (0.64, 0.89) and 0.72 (0.62, 0.84) h for ABCG2 421 genotypes CC, CA and AA, respectively. The percentage of dose excreted unchanged in the urine was 43, 44 and 39%, respectively. CONCLUSIONS: The ABCG2 C421A polymorphism had no effect on nitrofurantoin plasma and urine pharmacokinetic parameters in healthy Chinese subjects. These results indicate that nitrofurantoin is not a suitable clinical probe substrate for assessing BCRP activity.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Anti-Infective Agents, Urinary/pharmacokinetics , Asian People/genetics , Neoplasm Proteins/genetics , Nitrofurantoin/pharmacokinetics , Polymorphism, Genetic/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/genetics , Administration, Oral , Adult , Anti-Infective Agents, Urinary/administration & dosage , Biological Transport/drug effects , Drug Resistance, Neoplasm/genetics , Genotype , Humans , Male , Middle Aged , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Nitrofurantoin/administration & dosage , Treatment Outcome
13.
Regul Toxicol Pharmacol ; 49(2): 90-100, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17655994

ABSTRACT

To promptly identify and evaluate liver safety events, an evidence-based liver safety system was created for global Phase I-III clinical trials. The goals of this system included improving clinical trial subject safety, expanding information on liver safety events, and improving data quality across studies by establishing and communicating: Two different algorithms for liver stopping criteria were developed. The most stringent criteria were selected for healthy volunteers in Phase I studies, where no treatment benefit is anticipated and clinical safety data are limited. With an interest in assessing potential liver "tolerance" or adaptation with accruing safety information, slightly higher liver chemistry thresholds were set for Phase II-III studies. This paper will describe the importance of liver safety in drug development, laboratory tests used to monitor liver safety, the rationale for selected liver chemistry subject stopping criteria, and implementation of this safety system.


Subject(s)
Chemical and Drug Induced Liver Injury , Clinical Trials as Topic , Drug Monitoring/methods , Drug-Related Side Effects and Adverse Reactions , Evidence-Based Medicine , Liver/drug effects , Algorithms , Endpoint Determination , Humans , Liver Function Tests , Withholding Treatment
14.
Bioorg Med Chem Lett ; 17(16): 4670-7, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17560786

ABSTRACT

The design, synthesis, and SAR of a novel series of heterobiaryl phenethanolamine beta3 adrenergic receptor agonists are described. The furan analogue 49 was shown to elicit a significant dose-dependent lowering of plasma glucose in a rodent model of type 2 diabetes.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Binding Sites , Models, Molecular , Molecular Structure , Structure-Activity Relationship
15.
Br J Clin Pharmacol ; 62(3): 336-44, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16934050

ABSTRACT

AIMS: This study assessed the effects of the CYP3A inhibitors lopinavir/ritonavir (LPV/r) on the steady-state pharmacokinetics (PK) of aplaviroc (APL), a CYP3A4 substrate, in healthy subjects. METHODS: In Part 1, APL PK was determined in eight subjects who received a single oral 50-mg APL test dose with/without a single dose of 100 mg ritonavir (RTV). Part 2 was conducted as an open-label, single-sequence, three-period repeat dose study in a cohort of 24 subjects. Subjects received APL 400 mg every 12 h (b.i.d.) for 7 days (Period 1), LPV/r 400/100 mg b.i.d. for 14 days (Period 2) and APL 400 mg + LPV/r 400/100 mg b.i.d. for 7 days (Period 3). All doses were administered with a moderate fat meal. PK sampling occurred on day 7 of Periods 1 and 3 and day 14 of Period 2. RESULTS: In Part 1, a single RTV dose increased the APL AUC(0-infinity) by 2.1-fold [90% confidence interval (CI) 1.9, 2.4]. Repeat dose coadministration of APL with LPV/r increased APL exposures to a greater extent with the geometric least squares mean ratios (90% CI) being 7.7 (6.4, 9.3), 6.2 (4.8, 8.1) and 7.1 (5.6, 9.0) for the APL AUC, C(max), and C(min), respectively. No change in LPV AUC or C(max) and a small increase in RTV AUC and C(max) (28% and 32%) were observed. The combination of APL and LPV/r was well tolerated and adverse events were mild in severity with self-limiting gastrointestinal complaints most commonly reported. CONCLUSIONS: Coadministration of APL and LPV/r was well tolerated and resulted in significantly increased APL plasma concentrations.


Subject(s)
CCR5 Receptor Antagonists , HIV Infections/drug therapy , Pyrimidinones/pharmacology , Ritonavir/pharmacology , Adolescent , Adult , Animals , Benzoates/pharmacokinetics , Diketopiperazines , Drug Combinations , Drug Interactions , Female , Humans , Lopinavir , Male , Mice , Middle Aged , Piperazines/pharmacokinetics , Pyrimidinones/therapeutic use , Rats , Ritonavir/therapeutic use , Spiro Compounds/pharmacokinetics
16.
J Clin Pharmacol ; 46(5): 577-87, 2006 May.
Article in English | MEDLINE | ID: mdl-16638741

ABSTRACT

Aplaviroc is a novel CCR5 antagonist, a class of compounds under investigation as viral entry inhibitors for the treatment of human immunodeficiency virus infection. A modified Cooperstown 5+1 cocktail was used to assess the drug interaction potential of aplaviroc. Fifteen healthy subjects were administered single oral doses of caffeine (CYP1A2), warfarin (CYP2C9), omeprazole (CYP2C19), dextromethorphan (CYP2D6), and midazolam (CYP3A) alone (reference treatment) and during steady-state administration of aplaviroc (400 mg every 12 hours, test treatment). Metabolite-to-parent area under the plasma concentration versus time curve (AUC) ratios (paraxanthine/caffeine and 5-hydroxyomeprazole/omeprazole), oral clearance (S-warfarin), AUC (midazolam), and metabolite-to-parent urinary excretion ratio (dextrorphan/dextromethorphan) were determined. The test-to-reference treatment ratios (geometric mean ratio and 90% confidence interval) were caffeine, 1.06 (0.97-1.17); S-warfarin, 0.93 (0.76-1.15); omeprazole, 1.07 (0.98-1.16); dextromethorphan, 1.17 (0.97-1.42); midazolam, 1.30 (1.04-1.63). No significant inhibition of CYP1A2, CYP2C9, CYP2C19, or CYP2D6 enzyme activity was observed. Mild inhibition of CYP3A isozymes should not preclude the use of concomitant CYP3A substrates in future clinical studies with aplaviroc.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Benzoates/pharmacokinetics , Piperazines/pharmacokinetics , Spiro Compounds/pharmacokinetics , Adult , Anti-HIV Agents/adverse effects , Anti-HIV Agents/blood , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Benzoates/adverse effects , Benzoates/blood , CCR5 Receptor Antagonists , Caffeine/blood , Caffeine/pharmacokinetics , Dextromethorphan/pharmacokinetics , Dextromethorphan/urine , Diketopiperazines , Drug Interactions , Female , Humans , Male , Midazolam/blood , Midazolam/pharmacokinetics , Middle Aged , Omeprazole/blood , Omeprazole/pharmacokinetics , Piperazines/adverse effects , Piperazines/blood , Spiro Compounds/adverse effects , Spiro Compounds/blood , Warfarin/blood , Warfarin/pharmacokinetics
17.
Antimicrob Agents Chemother ; 49(7): 2802-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15980352

ABSTRACT

873140 is a novel CCR5 antagonist with potent in vitro anti-human immunodeficiency virus (HIV) activity. This study was a double-blind, randomized, placebo-controlled, single- and repeat-dose escalation investigation of the safety, pharmacokinetics, and food effect of 873140 in 70 adult subjects. During single-dose escalation, three cohorts (each composed of 10 subjects, with 8 subjects receiving the active drug and 2 subjects receiving the placebo [8 active and 2 placebo]) received doses of 50, 200, 400, 800, and 1,200 mg after an overnight fast, or 400 mg plus a standard high-fat breakfast in an alternating panel design. During repeat-dose escalation, four cohorts (each with 8 active and 2 placebo) received doses of 200, 400, 600, or 800 mg every 12 h (BID) for 8 days. Laboratory safety tests, vital signs, and electrocardiograms (ECGs) were performed at regular intervals, and blood samples were obtained for pharmacokinetics. Single and repeat doses of 50 mg to 800 mg were well tolerated, with no serious adverse events and no grade 3 or 4 adverse events. The mild-to-moderate side effects were primarily gastrointestinal and included abdominal cramping, nausea, and diarrhea. No specific trends in laboratory parameters or clinically significant ECG changes were noted. Plasma 873140 concentrations increased rapidly; the median time to maximum concentration of drug in serum was 1.75 to 5 h. The median area under the plasma concentration-time profile (AUC) and the maximum concentration of drug in serum (C(max)) ranged from 127 ng.h/ml and 24 ng/ml at 200 mg BID to 329 ng.h/ml and 100 ng/ml at 800 mg BID, respectively. Food consumption increased the AUC and C(max) by a mean of 1.7- and 2.2-fold, respectively. The pharmacokinetic and safety profile supports the continued investigation of 873140 with HIV-infected subjects.


Subject(s)
Benzoates , CCR5 Receptor Antagonists , HIV Fusion Inhibitors , Spiro Compounds , Adolescent , Adult , Benzoates/administration & dosage , Benzoates/adverse effects , Benzoates/metabolism , Benzoates/pharmacokinetics , Diketopiperazines , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Food-Drug Interactions , HIV Fusion Inhibitors/administration & dosage , HIV Fusion Inhibitors/adverse effects , HIV Fusion Inhibitors/metabolism , HIV Fusion Inhibitors/pharmacokinetics , Humans , Male , Middle Aged , Piperazines , Spiro Compounds/administration & dosage , Spiro Compounds/adverse effects , Spiro Compounds/metabolism , Spiro Compounds/pharmacokinetics
18.
Adv Drug Deliv Rev ; 56(12): 1825-57, 2004 Oct 14.
Article in English | MEDLINE | ID: mdl-15381336

ABSTRACT

Cerebrospinal fluid (CSF) concentrations are used as a surrogate measure of central nervous system (CNS) availability of drugs. Systemically administered drugs can reach CSF either directly via passage across the choroid plexus, or indirectly by passage across the blood-brain barrier (BBB) followed by diffusion/convection transport from the interstitial fluid (ISF) to CSF. This review focuses on the physiological and pharmacokinetic variables that must be considered in the interpretation of the CSF-to-plasma concentration gradient. A survey of the literature suggests that the equilibrated CSF-to-unbound plasma concentration ratio reflects the balance between drug permeability across the blood-CNS barriers and the sink action of CSF turnover. As lipophilicity and membrane permeability increase, the CSF-to-plasma unbound concentration ratio rises from well below 1 toward unity. Deviations are noted in that lipophilic drugs highly bound to CSF proteins had ratio exceeding unity, and lipophilic drugs that were efflux transporter substrates exhibited ratios well less than unity. Drug concentration gradients have been observed between drug in the ISF and CSF, the direction of which depends on the CSF sink action and drug permeability/transport across the BBB and choroid plexus. Despite the complexity of CSF pharmacokinetics, for some drugs a rapid kinetic equilibrium exists between the CSF and biophase, such that CSF concentration can serve as a proximate reference for detailed investigation of factors affecting the intrinsic pharmacodynamics of a centrally acting drug. The applicability and limitations of CSF sampling for assessing the CNS availability and concentration-effect relationship of drug candidates with varying physicochemical properties during drug discovery and development is discussed.


Subject(s)
Blood-Brain Barrier , Brain/metabolism , Cerebrospinal Fluid/metabolism , Animals , Biological Transport , Choroid Plexus/metabolism , Extracellular Fluid/metabolism , Humans
19.
Pharmacology ; 72(2): 92-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15331914

ABSTRACT

The purpose of this study was to measure the in vivo brain distribution of antihistamines and assess the influence of in vitro permeability, P-glycoprotein (Pgp) efflux, and plasma protein binding. Six antihistamines (acrivastine, chlorpheniramine, diphenhydramine doxylamine, fexofenadine, terfenadine) were selected based on previously reported in vitro permeability and Pgp efflux properties and dosed intravenously to steady-state plasma concentrations of 2-10 micromol/l in rats. Plasma and brain concentrations were measured by LC/MS/MS, and protein binding determined by ultrafiltration. Doxylamine, diphenhydramine and chlorpheniramine had brain-to-plasma concentration ratios of 4.34 +/- 1.26, 18.4 +/- 2.35 and 34.0 +/- 9.02, respectively. These drugs had high passive membrane permeability (>310 nm/s), moderate protein binding (71-84%) and were not Pgp substrates; features that yield high CNS penetration. In contrast, acrivastine and fexofenadine had low brain-to-plasma ratios of 0.072 +/- 0.014 and 0.018 + 0.002, consistent with low passive membrane permeability for both compounds (16.2 and 66 nm/s, respectively) and Pgp efflux. Finally, terfenadine had a brain-to-plasma ratio of 2.21 +/- 1.00 even though it underwent Pgp-mediated efflux (in vitro ratio = 2.88). Terfenadine's high passive permeability (285 nm/s) overcame the Pgp-mediated efflux to yield brain-to-plasma ratio >1. The brain-to-unbound plasma ratio was 22-fold higher suggesting that protein binding (96.3% bound) limited terfenadine's brain distribution. In conclusion, passive membrane permeability, Pgp-mediated efflux and/or high plasma protein binding influence the in vivo brain distribution of antihistamine drugs.


Subject(s)
Brain/metabolism , Histamine H1 Antagonists/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Blood Proteins/metabolism , Cell Membrane Permeability , Male , Protein Binding , Rats , Rats, Wistar , Tissue Distribution
20.
J Pharmacol Exp Ther ; 303(3): 1029-37, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12438524

ABSTRACT

Membrane permeability and P-glycoprotein (Pgp) can be limiting factors for blood-brain barrier penetration. The objectives of this study were to determine whether there are differences in the in vitro permeability, Pgp substrate profiles, and physicochemical properties of drugs for central nervous system (CNS) and non-CNS indications, and whether these differences are useful criteria in selecting compounds for drug development. Apparent permeability (P(app)) and Pgp substrate profiles for 93 CNS (n = 48) and non-CNS (n = 45) drugs were determined by monolayer efflux. Calcein-AM inhibition assays were used to supplement the efflux results. The CNS set (2 of 48, 4.2%) had a 7-fold lower incidence of passive permeability values <150 nm/s compared with the non-CNS set (13 of 45, 28.9%). The majority of drugs (72.0%, 67 of 93) were not Pgp substrates; however, 49.5% (46 of 93) were positive in the calcein-AM assay when tested at 100 microM. The CNS drug set (n = 7 of 48, 14.6%) had a 3-fold lower incidence of Pgp-mediated efflux than the non-CNS drug set (n = 19 of 45, 42.2%). Analysis of 18 physicochemical properties revealed that the CNS drug set had fewer hydrogen bond donors, fewer positive charges, greater lipophilicity, lower polar surface area, and reduced flexibility compared with the non-CNS group (p < 0.05), properties that enhance membrane permeability. This study on a large, diverse set of marketed compounds clearly demonstrates that permeability, Pgp-mediated efflux, and certain physicochemical properties are factors that differentiate CNS and non-CNS drugs. For CNS delivery, a drug should ideally have an in vitro passive permeability >150 nm/s and not be a good (B --> A/A --> B ratio <2.5) Pgp substrate.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Blood-Brain Barrier/drug effects , Cell Membrane Permeability/drug effects , Central Nervous System Agents/pharmacokinetics , Animals , Blood-Brain Barrier/physiology , Cell Line , Central Nervous System Agents/pharmacology , Dogs , Drug Delivery Systems/methods , Permeability/drug effects , Pharmaceutical Preparations/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...